1
|
Félix P, Melo AA, Costa JP, Colaço M, Pereira D, Núñez J, de Almeida LP, Borges O. Exploring TLR agonists as adjuvants for COVID-19 oral vaccines. Vaccine 2025; 53:127078. [PMID: 40184639 DOI: 10.1016/j.vaccine.2025.127078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/18/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025]
Abstract
The COVID-19 pandemic underscored the importance of advancing technologies that enable the rapid development and distribution of more effective vaccines when required. Since SARS-CoV-2 enters the body through the nasal mucosa, optimising the induction of secretory IgA (sIgA) production, a key component of the mucosal immune response, is essential. It has long been known that the induction of sIgA occurs when a vaccine is administered through mucosal surfaces and the immune responses initiated at one mucosal site can influence immune activity at other mucosal surfaces. Consequently, we propose an oral vaccine formulation (Vacform) comprising the immunomodulator CL097, a TLR7/8 agonist, and the SARS-CoV-2 spike protein, both encapsulated within glucan particles (GPs). The studies demonstrated that Vacform induced ROS production in RAW 264.7 cells but not in human neutrophils. The concentrations of Vacform tested did not induce NO production in RAW 264.7 cells. While Vacform stimulated the production of TNF-α and IL-6 in mouse spleen cells, this effect was not observed in RAW 264.7 cells. Finally, Vacform stimulated the proliferation of human PBMCs. Thus, its immunomodulatory properties were evident in specific cells under certain in vitro conditions. The Vacform was subsequently tested in vaccination studies. C57BL/6 mice were initially immunized subcutaneously, followed by two oral boosts with Vacform every two weeks. The Vacform elicited both, humoral (serum IgG and mucosal sIgA) and cellular immune responses. A balanced Th1/Th2/Th17 immune profile was observed. In conclusion, the GPs:CL097 adjuvant system shows promise for eliciting robust immune responses against SARS-CoV-2 and provides a foundation for future studies on dose-response optimization and challenge models.
Collapse
Affiliation(s)
- Paulo Félix
- Faculty of Pharmacy (FFUC), University of Coimbra, 3000-548 Coimbra, Portugal; Centre for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra 3004-504, Portugal; Centre for Neuroscience and Cell Biology - University of Coimbra (CNC - UC), Coimbra 3004-504, Portugal
| | - Alexandra A Melo
- Faculty of Pharmacy (FFUC), University of Coimbra, 3000-548 Coimbra, Portugal; Centre for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra 3004-504, Portugal; Centre for Neuroscience and Cell Biology - University of Coimbra (CNC - UC), Coimbra 3004-504, Portugal
| | - João Panão Costa
- Faculty of Pharmacy (FFUC), University of Coimbra, 3000-548 Coimbra, Portugal; Centre for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra 3004-504, Portugal; Centre for Neuroscience and Cell Biology - University of Coimbra (CNC - UC), Coimbra 3004-504, Portugal
| | - Mariana Colaço
- Faculty of Pharmacy (FFUC), University of Coimbra, 3000-548 Coimbra, Portugal; Centre for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra 3004-504, Portugal; Centre for Neuroscience and Cell Biology - University of Coimbra (CNC - UC), Coimbra 3004-504, Portugal
| | - Dina Pereira
- Centre for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra 3004-504, Portugal; Centre for Neuroscience and Cell Biology - University of Coimbra (CNC - UC), Coimbra 3004-504, Portugal; Gene Therapy Center of Excellence (GeneT), Coimbra 3004-504, Portugal; IIIUC - Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Jisette Núñez
- Faculty of Pharmacy (FFUC), University of Coimbra, 3000-548 Coimbra, Portugal; Centre for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra 3004-504, Portugal; Centre for Neuroscience and Cell Biology - University of Coimbra (CNC - UC), Coimbra 3004-504, Portugal; Gene Therapy Center of Excellence (GeneT), Coimbra 3004-504, Portugal
| | - Luís Pereira de Almeida
- Faculty of Pharmacy (FFUC), University of Coimbra, 3000-548 Coimbra, Portugal; Centre for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra 3004-504, Portugal; Centre for Neuroscience and Cell Biology - University of Coimbra (CNC - UC), Coimbra 3004-504, Portugal; Gene Therapy Center of Excellence (GeneT), Coimbra 3004-504, Portugal
| | - Olga Borges
- Faculty of Pharmacy (FFUC), University of Coimbra, 3000-548 Coimbra, Portugal; Centre for Innovation in Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra 3004-504, Portugal; Centre for Neuroscience and Cell Biology - University of Coimbra (CNC - UC), Coimbra 3004-504, Portugal.
| |
Collapse
|
2
|
Hygino J, Sales MC, Sacramento PM, Kasahara TM, da Silva JCC, Bilhão R, Andrade RM, Vasconcelos CCF, Bento CAM. Hyperresponsiveness of Corticoid-Resistant Th17/Tc-17 Cells to TLR-2 and TLR-4 Ligands is a Feature of Multiple Sclerosis Patients at Higher Risk of Therapy Failure. J Inflamm Res 2024; 17:8775-8797. [PMID: 39564547 PMCID: PMC11573880 DOI: 10.2147/jir.s476110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 09/25/2024] [Indexed: 11/21/2024] Open
Abstract
Purpose The presence of T cells expressing TLR-2 and TLR-4 has been associated with relapsing-remitting multiple sclerosis (RRMS) pathogenesis. Here, we evaluated whether the effectiveness of DMT in controlling clinical activity of the disease would be associated with modulation of proportion of TLRs+ T cells. Patients and Methods Whole peripheral blood mononuclear cells, purified CD4+ and CD8+ T cells from RRMS patients were cultured with different stimuli. The frequency of IL-17-secreting CD4+ and CD8+ T cells positive for TLR-2 and TLR-4 was determined by flow cytometry. The cytokine profile of these T cells following TLR-2 and TLR-4 stimulation was determined by Multiplex. Some of these T cell cultures were treated with hydrocortisone. The levels of LPS-binding protein (LBP) were dosed by ELISA. Clinical (occurrence of relapses) and radiological (number of active brain lesions) activity were evaluated during the 1-year follow-up. Results Despite DMT, high intensity of TLR-2 and TLR-4 expression on (CD4+ and CD8+) T-cells, as well as the frequency of IL-17-secreting (CD4+ and CD8+) T-cells, are predictive of future RRMS relapses. Moreover, higher cytokine production related to Th17/Tc-17 phenotypes in response to TLR-2 and TLR-4 agonists was observed in DMT-treated patients and displayed an elevated number of brain lesions. The hyperresponsiveness of MS-derived T-cells to TLR-2 and TLR-4 ligands, with high levels of IL-1β, IL-6, IL-17, IFN-γ and GM-CSF in response to both TLR agonists, positively correlated with plasma LBP levels. Interestingly, corticoid was less efficient in reducing Th17 and Tc-17 cytokine production induced by TLR-2 and TLR-4 ligands in DMT-treated patients who relapsed during follow-up. Conclusion Collectively, the data suggested that persistence of circulating Th17 and Tc17 cells expressing elevated levels of functional TLR-2 and TLR-4 could indicate high disease activity and lower therapeutic efficacy in RRMS patients.
Collapse
Affiliation(s)
- Joana Hygino
- Post-Graduate Program in Neurology, Federal University of the State of Rio de Janeiro, Rio de Janeiro City, Brazil
| | - Marisa C Sales
- Post-Graduate Program in Microbiology, University of the State of Rio de Janeiro, Rio de Janeiro City, Brazil
| | - Priscila M Sacramento
- Post-Graduate Program in Neurology, Federal University of the State of Rio de Janeiro, Rio de Janeiro City, Brazil
| | - Taissa M Kasahara
- Post-graduate Program in Molecular and Cellular Biology, Federal University of the State of Rio de Janeiro, Rio de Janeiro City, Brazil
| | - Júlio César Costa da Silva
- Post-graduate Program in Molecular and Cellular Biology, Federal University of the State of Rio de Janeiro, Rio de Janeiro City, Brazil
| | - Rafaela Bilhão
- Post-graduate Program in Molecular and Cellular Biology, Federal University of the State of Rio de Janeiro, Rio de Janeiro City, Brazil
| | - Regis M Andrade
- Department of General Medicine, Federal University of the State of Rio de Janeiro, Rio de Janeiro City, Brazil
| | | | - Cleonice A M Bento
- Post-Graduate Program in Neurology, Federal University of the State of Rio de Janeiro, Rio de Janeiro City, Brazil
- Post-graduate Program in Molecular and Cellular Biology, Federal University of the State of Rio de Janeiro, Rio de Janeiro City, Brazil
- Department of General Medicine, Federal University of the State of Rio de Janeiro, Rio de Janeiro City, Brazil
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro City, Brazil
| |
Collapse
|
3
|
Newman MJ. Invention and characterization of a systemically administered, attenuated and killed bacteria-based multiple immune receptor agonist for anti-tumor immunotherapy. Front Immunol 2024; 15:1462221. [PMID: 39606250 PMCID: PMC11599860 DOI: 10.3389/fimmu.2024.1462221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/18/2024] [Indexed: 11/29/2024] Open
Abstract
Activation of immune receptors, such as Toll-like (TLR), NOD-like (NLR) and Stimulator of Interferon Genes (STING) is critical for efficient innate and adaptive immunity. Gram-negative bacteria (G-NB) contain multiple TLR, NOD and STING agonists. Potential utility of G-NB for cancer immunotherapy is supported by observations of tumor regression in the setting of infection and Coley's Toxins. Coley reported that intravenous (i.v.) administration was likely most effective but produced uncontrollable toxicity. The discovery of TLRs and their agonists, particularly the potent TLR4 agonist lipopolysaccharide (LPS)-endotoxin, comprising ~75% of the outer membrane of G-NB, suggests that LPS may be both a critical active ingredient and responsible for dose-limiting i.v. toxicity of G-NB. This communication reports the production of killed, stabilized, intact bacteria products from non-pathogenic G-NB with ~96% reduction of LPS-endotoxin activity. One resulting product candidate, Decoy10, was resistant to standard methods of cell disruption and contained TLR2,4,8,9, NOD2 and STING agonist activity. Decoy10 also exhibited reduced i.v. toxicity in mice and rabbits, and a largely uncompromised ability to induce cytokine and chemokine secretion by human immune cells in vitro, all relative to unprocessed, parental bacterial cells. Decoy10 and a closely related product, Decoy20, produced single agent anti-tumor activity or combination-mediated durable regression of established subcutaneous, metastatic or orthotopic colorectal, hepatocellular (HCC), pancreatic, and non-Hodgkin's lymphoma (NHL) tumors in mice, with induction of both innate and adaptive immunological memory (syngeneic and human tumor xenograft models). Decoy bacteria combination-mediated regressions were observed with a low-dose, oral non-steroidal anti-inflammatory drug (NSAID), anti-PD-1 checkpoint therapy, low-dose cyclophosphamide (LDC), and/or a targeted antibody (rituximab). Efficient tumor eradication was associated with plasma expression of 15-23 cytokines and chemokines, broad induction of cytokine, chemokine, innate and adaptive immune pathway genes in tumors, cold to hot tumor inflammation signature transition, and required NK, CD4+ and CD8+ T cells, collectively demonstrating a role for both innate and adaptive immune activation in the anti-tumor immune response.
Collapse
|
4
|
Uddin MB, Roy KR, Hill RA, Roy SC, Gu X, Li L, Zhang QJ, You Z, Liu YY. p53 missense mutant G242A subverts natural killer cells in sheltering mouse breast cancer cells against immune rejection. Exp Cell Res 2022; 417:113210. [PMID: 35597298 DOI: 10.1016/j.yexcr.2022.113210] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 05/07/2022] [Accepted: 05/14/2022] [Indexed: 11/15/2022]
Abstract
Cancer cells acquire immunoediting ability to evade immune surveillance and thus escape eradication. It is widely known that mutant proteins encoded from tumor suppressor TP53 exhibit gain-of-function in cancer cells, thereby promoting progression; however, how mutant p53 contributes to the sheltering of cancer cells from host anticancer immunity remains unclear. Herein, we report that murine p53 missense mutation G242A (corresponding to human G245A) suppresses the activation of host natural killer (NK) cells, thereby enabling breast cancer cells to avoid immune assault. We found that serial injection of EMT6 breast cancer cells that carry wild-type (wt) Trp53, like normal fibroblasts, promoted NK activity in mice, while SVTneg2 cells carrying Trp53 G242A+/+ mutation decreased NK cell numbers and increased CD8+ T lymphocyte numbers in spleen. Innate immunity based on NK cells and CD8 T cells was reduced in p53 mutant-carrying transgenic mice (Trp53 R172H/+, corresponding to human R175H/+). Further, upon co-culture with isolated NK cells, EMT6 cells substantively activated NK cells and proliferation thereof, increasing interferon-gamma (IFN-γ) production; however, SVTneg2 cells suppressed NK cell activation. Further mechanistic study elucidated that p53 can modulate expression by cancer cells of Mult-1 and H60a, which are activating and inhibitory ligands for NKG2D receptors of NK cells, respectively, to enhance immune surveillance against cancer. Our findings demonstrate that wt p53 is requisite for NK cell-based immune recognition and elimination of cancerous cells, and perhaps more importantly, that p53 missense mutant presence in cancer cells impairs NK cell-attributable responses, thus veiling cancerous cells from host immunity and enabling cancer progression.
Collapse
Affiliation(s)
- Mohammad B Uddin
- School of Basic Pharmaceutical and Toxicological Sciences, University of Louisiana at Monroe, Monroe, LA, USA
| | - Kartik R Roy
- School of Basic Pharmaceutical and Toxicological Sciences, University of Louisiana at Monroe, Monroe, LA, USA
| | - Ronald A Hill
- School of Basic Pharmaceutical and Toxicological Sciences, University of Louisiana at Monroe, Monroe, LA, USA
| | - Sagor C Roy
- School of Basic Pharmaceutical and Toxicological Sciences, University of Louisiana at Monroe, Monroe, LA, USA
| | - Xin Gu
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Li Li
- Laboratory of Translational Cancer Research, Tom & Gayle Benson Cancer Center, Ochsner Clinic Foundation, New Orleans, LA, USA
| | - Qian-Jin Zhang
- Department of Biology, Xavier University of Louisiana, New Orleans, LA, USA
| | - Zongbing You
- Department of Structural and Cellular Biology, Tulane University, New Orleans, LA, USA
| | - Yong-Yu Liu
- School of Basic Pharmaceutical and Toxicological Sciences, University of Louisiana at Monroe, Monroe, LA, USA.
| |
Collapse
|
5
|
Castro C, Oyamada HAA, Cafasso MOSD, Lopes LM, Monteiro C, Sacramento PM, Alves-Leon SV, da Fontoura Galvão G, Hygino J, de Souza JPBM, Bento CAM. Elevated proportion of TLR2- and TLR4-expressing Th17-like cells and activated memory B cells was associated with clinical activity of cerebral cavernous malformations. J Neuroinflammation 2022; 19:28. [PMID: 35109870 PMCID: PMC8808981 DOI: 10.1186/s12974-022-02385-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 01/12/2022] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Recent evidences have suggested the involvement of toll-like receptor (TLR)-4 in the pathogenesis of cerebral cavernous malformations (CCM). Elevated frequency of TLR+T-cells has been associated with neurological inflammatory disorders. As T-cells and B-cells are found in CCM lesions, the objective of the present study was to evaluate the cytokine profile of T-cells expressing TLR2 and TLR4, as well as B-cell subsets, in asymptomatic (CCMAsympt) and symptomatic (CCMSympt) patients. METHODS For our study, the cytokine profile from TLR2+ and TLR4+ T-cell and B-cell subsets in CCMAsympt and CCMSympt patients was investigated using flow cytometry and ELISA. T-cells were stimulated in vitro with anti-CD3/anti-CD28 beads or TLR2 (Pam3C) and TLR4 (LPS) ligands. RESULTS CCMSymptc patients presented a higher frequency of TLR4+(CD4+ and CD8+) T-cells and greater density of TLR4 expression on these cells. With regard to the cytokine profile, the percentage of TLR2+ and TLR4+ Th17 cells was higher in CCMSympt patients. In addition, an elevated proportion of TLR4+ Tc-1 cells, as well as Tc-17 and Th17.1 cells expressing TLR2 and TLR4, was observed in the symptomatic patients. By contrast, the percentage of TLR4+ IL-10+CD4+ T cells was higher in the CCMAsympt group. Both Pam3C and LPS were more able to elevate the frequency of IL-6+CD4+T cells and Th17.1 cells in CCMSympt cell cultures. Furthermore, in comparison with asymptomatic patients, purified T-cells from the CCMSympt group released higher levels of Th17-related cytokines in response to Pam3C and, mainly, LPS, as well as after activation via TCR/CD28. Concerning the B-cell subsets, a higher frequency of memory and memory activated B-cells was observed in CCMSympt patients. CONCLUSIONS Our findings reveal an increase in circulating Th17/Tc-17 cell subsets expressing functional TLR2 and, mainly, TLR4 molecules, associated with an increase in memory B-cell subsets in CCM patients with clinical activity of the disease.
Collapse
Affiliation(s)
- Camilla Castro
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Frei Caneca 94, Rio de Janeiro, RJ, 20261-040, Brazil
- Post-Graduate Program in Microbiology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Hugo A A Oyamada
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Frei Caneca 94, Rio de Janeiro, RJ, 20261-040, Brazil
- Post-Graduate Program in Microbiology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcos Octávio S D Cafasso
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Frei Caneca 94, Rio de Janeiro, RJ, 20261-040, Brazil
| | - Lana M Lopes
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Frei Caneca 94, Rio de Janeiro, RJ, 20261-040, Brazil
- Post-Graduate Program in Microbiology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Clarice Monteiro
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Frei Caneca 94, Rio de Janeiro, RJ, 20261-040, Brazil
- Post-Graduate Program in Microbiology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Priscila M Sacramento
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Frei Caneca 94, Rio de Janeiro, RJ, 20261-040, Brazil
- Post-Graduate Program in Microbiology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Soniza Vieira Alves-Leon
- Post-Graduate Program in Neurology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
- Translational Neuroscience Laboratory (LabNet), University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gustavo da Fontoura Galvão
- Service of Neurosurgery, University Hospital of the Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Joana Hygino
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Frei Caneca 94, Rio de Janeiro, RJ, 20261-040, Brazil
- Post-Graduate Program in Neurology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jorge Paes Barreto Marcondes de Souza
- Post-Graduate Program in Neurology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
- Service of Neurosurgery, University Hospital of the Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Post-Graduate Program of Surgical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cleonice A M Bento
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Frei Caneca 94, Rio de Janeiro, RJ, 20261-040, Brazil.
- Post-Graduate Program in Microbiology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
- Post-Graduate Program in Neurology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
6
|
Sales MC, Kasahara TM, Sacramento PM, Rossi ÁD, Cafasso MOS, Oyamada HA, Hygino J, Alvim F, Andrade RM, Cristina Vasconcelos C, Bento CA. Selective serotonin reuptake inhibitor attenuates the hyperresponsiveness of TLR2 + and TLR4 + Th17/Tc17-like cells in multiple sclerosis patients with major depression. Immunology 2021; 162:290-305. [PMID: 33112414 PMCID: PMC7884649 DOI: 10.1111/imm.13281] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/18/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023] Open
Abstract
Elevated frequency of Th17-like cells expressing Toll-like receptors (TLRs) has been recently associated with relapsing-remitting multiple sclerosis (MS) pathogenesis, a chronic inflammatory demyelinating autoimmune disease of the central nervous system. We aimed to investigate the impact of current major depressive disorder (MDD) on the behaviour of these cells following in vitro stimulation with TLR2, TLR4, TLR5 and TLR9 agonists. Here, the level of both cell proliferation and cytokine production related to Th17/Tc17 phenotypes in response to TLR2 (Pam3C) and TLR4 (LPS) ligands was significantly higher in CD4+ and CD8+ T-cell cultures from MS/MDD patients when compared to non-depressed patients. These cytokine levels were positively associated with neurological disabilities in patients. No difference for responsiveness to TLR5 (flagellin) and TLR9 (ODN) agonists was observed. LPS, but not Pam3C, induced significant IL-10 release, mainly in patients without MDD. Interestingly, more intense expression of TLR2 and TLR4 on these cells was observed in MDD patients. Finally, in vitro addition of serotonin and treatment of MDD patients with selective serotonin reuptake inhibitors (SSRIs) reduced the production of Th17/Tc17-related cytokines by CD4+ and CD8+ T cells in response to Pam3C and LPS. However, only SSRI therapy diminished the frequency and intensity of TLR2 and TLR4 expression on circulating CD4+ and CD8+ T cells. In summary, although preliminary, our findings suggest that adverse events that elevate circulating levels of TLR2 and TLR4 ligands can affect MS pathogenesis, particularly among depressed patients.
Collapse
Affiliation(s)
- Marisa C. Sales
- Department of Microbiology and ParasitologyFederal University of the State of Rio de JaneiroRio de JaneiroBrazil
- Post‐graduate Program in MicrobiologyUniversity of the State of Rio de JaneiroRio de JaneiroBrazil
| | - Taissa M. Kasahara
- Department of Microbiology and ParasitologyFederal University of the State of Rio de JaneiroRio de JaneiroBrazil
| | - Priscila M. Sacramento
- Department of Microbiology and ParasitologyFederal University of the State of Rio de JaneiroRio de JaneiroBrazil
- Post‐graduate Program in MicrobiologyUniversity of the State of Rio de JaneiroRio de JaneiroBrazil
| | - Átila D. Rossi
- Department of GeneticsFederal University of Rio de JaneiroRio de JaneiroBrazil
| | - Marcos Octávio S.D. Cafasso
- Department of Microbiology and ParasitologyFederal University of the State of Rio de JaneiroRio de JaneiroBrazil
| | - Hugo A.A. Oyamada
- Department of Microbiology and ParasitologyFederal University of the State of Rio de JaneiroRio de JaneiroBrazil
- Post‐graduate Program in MicrobiologyUniversity of the State of Rio de JaneiroRio de JaneiroBrazil
| | - Joana Hygino
- Department of Microbiology and ParasitologyFederal University of the State of Rio de JaneiroRio de JaneiroBrazil
- Post‐graduate Program in NeurologyFederal University of the State of Rio de JaneiroRio de JaneiroBrazil
| | - Fabianna Alvim
- Department of Microbiology and ParasitologyFederal University of the State of Rio de JaneiroRio de JaneiroBrazil
| | - Regis M. Andrade
- Department of General Medicine DepartmentFederal University of the State of Rio de JaneiroRio de JaneiroBrazil
| | | | - Cleonice A.M. Bento
- Department of Microbiology and ParasitologyFederal University of the State of Rio de JaneiroRio de JaneiroBrazil
- Post‐graduate Program in NeurologyFederal University of the State of Rio de JaneiroRio de JaneiroBrazil
- Department of General Medicine DepartmentFederal University of the State of Rio de JaneiroRio de JaneiroBrazil
| |
Collapse
|
7
|
Glucose metabolism characteristics and TLR8-mediated metabolic control of CD4 + Treg cells in ovarian cancer cells microenvironment. Cell Death Dis 2021; 12:22. [PMID: 33414414 PMCID: PMC7790820 DOI: 10.1038/s41419-020-03272-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 12/13/2022]
Abstract
Immunotherapy is expected to become the most promising new treatment for ovarian cancer owing to its immunogenicity. However, immunosuppression in the tumor microenvironment is a major obstacle to the efficacy of tumor therapy. Studies have found different metabolism ways of regulatory T cells (Tregs) in the cancer environment may be related to the immunosuppression and Toll-like receptor 8 (TLR8) can reverse the suppression function of Tregs. But it is still unclear that if the TLR8-mediated function reversal is associated with the change of glucose metabolism of Tregs. It was found that the positive expression rates of Glut1, HIF-1α, and Ki67 in CD4+ Treg cells of OC were significantly higher than that in benign ovarian tumor and HC, and also significantly higher than that in CD4+ Teffs of OC. What’s more, compared with CD4+ Teff group, CD4+ Tregs highly expressed seven genes and three proteins related to glucose metabolism and had higher levels of glucose uptake and glycolysis. After activating TLR8 signal of CD4+ Tregs, the proliferation level of naive CD4+ T cells was higher than that of the control group. At the same time, the expression levels of eight genes and five proteins related to glucose metabolism in CD4+ Treg cells with TLR8 activated were decreased and levels of glucose uptake and glycolysis were also lower. Furthermore, TLR8 signaling also downregulated the mTOR pathway in CD4+ Tregs. CD4+ Tregs pretreated with 2-deoxy-d-Glucose (2-DG) and galloflavin also attenuated the inhibition of Teffs proliferation. Although CD4+ Tregs pretreated with 2-DG and galloflavin before activating TLR8 signal had no significant difference compared with the group only treated with inhibitors, which suggested TLR8-mediated reversal of CD4+ Treg cells inhibitory function in ovarian cancer cells co-cultured microenvironment had a causal relationship with glucose metabolism.
Collapse
|
8
|
Nakao M, Sugaya M, Fujita H, Miyagaki T, Morimura S, Shibata S, Asano Y, Sato S. TLR2 Deficiency Exacerbates Imiquimod-Induced Psoriasis-Like Skin Inflammation through Decrease in Regulatory T Cells and Impaired IL-10 Production. Int J Mol Sci 2020; 21:E8560. [PMID: 33202847 PMCID: PMC7696365 DOI: 10.3390/ijms21228560] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/07/2020] [Accepted: 11/10/2020] [Indexed: 12/30/2022] Open
Abstract
Emerging evidence has demonstrated that Toll-like receptors (TLRs) are associated with autoimmune diseases. In this study, we investigated the role of TLR2 in psoriasis using imiquimod-induced psoriasis-like dermatitis. Although TLR2 signaling is known to play a critical role in the induction of proinflammatory cytokines by immune cells, such as dendritic cells (DCs), macrophages, and monocytes, TLR2 deficiency unexpectedly exacerbated psoriasiform skin inflammation. Importantly, messenger RNA (mRNA) levels of Foxp-3 and IL-10 in the lesional skin were significantly decreased in TLR2 KO mice compared with wild-type mice. Furthermore, flow cytometric analysis of the lymph nodes revealed that the frequency of regulatory T cells (Tregs) among CD4-positive cells was decreased. Notably, stimulation with Pam3CSK4 (TLR2/1 ligand) or Pam2CSK4 (TLR2/6 ligand) increased IL-10 production from Tregs and DCs and the proliferation of Tregs. Finally, adoptive transfer of Tregs from wild-type mice reduced imiquimod-induced skin inflammation in TLR2 KO mice. Taken together, our results suggest that TLR2 signaling directly enhances Treg proliferation and IL-10 production by Tregs and DCs, suppressing imiquimod-induced psoriasis-like skin inflammation. Enhancement of TLR2 signaling may be a new therapeutic strategy for psoriasis.
Collapse
Affiliation(s)
- Momoko Nakao
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan; (M.N.); (H.F.); (T.M.); (S.M.); (S.S.); (Y.A.); (S.S.)
| | - Makoto Sugaya
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan; (M.N.); (H.F.); (T.M.); (S.M.); (S.S.); (Y.A.); (S.S.)
- Department of Dermatology, International University of Health and Welfare, Chiba 286-8520, Japan
| | - Hideki Fujita
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan; (M.N.); (H.F.); (T.M.); (S.M.); (S.S.); (Y.A.); (S.S.)
- Division of Cutaneous Science, Department of Dermatology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Tomomitsu Miyagaki
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan; (M.N.); (H.F.); (T.M.); (S.M.); (S.S.); (Y.A.); (S.S.)
- Department of Dermatology, St. Marianna University School of Medicine, Kanagawa 216-8511, Japan
| | - Sohshi Morimura
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan; (M.N.); (H.F.); (T.M.); (S.M.); (S.S.); (Y.A.); (S.S.)
- Department of Dermatology, International University of Health and Welfare, Chiba 286-8520, Japan
| | - Sayaka Shibata
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan; (M.N.); (H.F.); (T.M.); (S.M.); (S.S.); (Y.A.); (S.S.)
| | - Yoshihide Asano
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan; (M.N.); (H.F.); (T.M.); (S.M.); (S.S.); (Y.A.); (S.S.)
| | - Shinichi Sato
- Department of Dermatology, The University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan; (M.N.); (H.F.); (T.M.); (S.M.); (S.S.); (Y.A.); (S.S.)
| |
Collapse
|
9
|
Abstract
Personalized cancer vaccines (PCVs) are reinvigorating vaccine strategies in cancer immunotherapy. In contrast to adoptive T-cell therapy and checkpoint blockade, the PCV strategy modulates the innate and adaptive immune systems with broader activation to redeploy antitumor immunity with individualized tumor-specific antigens (neoantigens). Following a sequential scheme of tumor biopsy, mutation analysis, and epitope prediction, the administration of neoantigens with synthetic long peptide (SLP) or mRNA formulations dramatically improves the population and activity of antigen-specific CD4+ and CD8+ T cells. Despite the promising prospect of PCVs, there is still great potential for optimizing prevaccination procedures and vaccine potency. In particular, the arduous development of tumor-associated antigen (TAA)-based vaccines provides valuable experience and rational principles for augmenting vaccine potency which is expected to advance PCV through the design of adjuvants, delivery systems, and immunosuppressive tumor microenvironment (TME) reversion since current personalized vaccination simply admixes antigens with adjuvants. Considering the broader application of TAA-based vaccine design, these two strategies complement each other and can lead to both personalized and universal therapeutic methods. Chemical strategies provide vast opportunities for (1) exploring novel adjuvants, including synthetic molecules and materials with optimizable activity, (2) constructing efficient and precise delivery systems to avoid systemic diffusion, improve biosafety, target secondary lymphoid organs, and enhance antigen presentation, and (3) combining bioengineering methods to innovate improvements in conventional vaccination, "smartly" re-educate the TME, and modulate antitumor immunity. As chemical strategies have proven versatility, reliability, and universality in the design of T cell- and B cell-based antitumor vaccines, the union of such numerous chemical methods in vaccine construction is expected to provide new vigor and vitality in cancer treatment.
Collapse
Affiliation(s)
- Wen-Hao Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, 100084 Beijing, China
| | - Yan-Mei Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, 100084 Beijing, China.,Beijing Institute for Brain Disorders, 100069 Beijing, China.,Center for Synthetic and Systems Biology, Tsinghua University, 100084 Beijing, China
| |
Collapse
|
10
|
Regulatory B cells in infection, inflammation, and autoimmunity. Cell Immunol 2020; 352:104076. [PMID: 32143836 DOI: 10.1016/j.cellimm.2020.104076] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 02/18/2020] [Accepted: 02/20/2020] [Indexed: 12/14/2022]
Abstract
Regulatory B (Breg) cells are characterized by differential expression of CD5 and CD1d in mouse and CD24 and CD38 in human immune systems. The Breg family also includes LAG-3+CD138hi plasma cells, CD1d CD5 CD21 CD23 cells, Tim1, PD-L1, PD-L2, CD200- expressing B cells, and CD39hiKi67+ cells originating from the transitional, marginal zone or germinal centre of the spleen. Breg cells produce IL10 and IL35 and to cause immunosuppression. These cells respond to TLR2, TLR4, and TLR9 agonists, CD40 ligands, IL12p35 and heat shock proteins. Emerging evidence suggests that TLR signalling component Myd88 impacts the modulation of Breg cell responses and the host's susceptibility to infection. Breg cells are found to reduce relapsing-remitting experimental autoimmune encephalomyelitis. However, the Breg-mediated mechanism used to control T cell-mediated immune responses is still unclear. Here, we review the existing literature to find gaps in the current knowledge and to build a pathway to further research.
Collapse
|
11
|
Pesce S, Greppi M, Ferretti E, Obino V, Carlomagno S, Rutigliani M, Thoren FB, Sivori S, Castagnola P, Candiani S, Marcenaro E. miRNAs in NK Cell-Based Immune Responses and Cancer Immunotherapy. Front Cell Dev Biol 2020; 8:119. [PMID: 32161759 PMCID: PMC7053181 DOI: 10.3389/fcell.2020.00119] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 02/11/2020] [Indexed: 12/16/2022] Open
Abstract
The incidence of certain forms of tumors has increased progressively in recent years and is expected to continue growing as life expectancy continues to increase. Tumor-infiltrating NK cells may contribute to develop an anti-tumor response. Optimized combinations of different cancer therapies, including NK cell-based approaches for targeting tumor cells, have the potential to open new avenues in cancer immunotherapy. Functional inhibitory receptors on NK cells are needed to prevent their attack on healthy cells. Nevertheless, disruption of inhibitory receptors function on NK cells increases the cytotoxic capacity of NK cells against cancer cells. MicroRNAs (miRNAs) are small non-coding RNA molecules that target mRNA and thus regulate the expression of genes involved in the development, maturation, and effector functions of NK cells. Therapeutic strategies that target the regulatory effects of miRNAs have the potential to improve the efficiency of cancer immunotherapy. Interestingly, emerging evidence points out that some miRNAs can, directly and indirectly, control the surface expression of immune checkpoints on NK cells or that of their ligands on tumor cells. This suggests a possible use of miRNAs in the context of anti-tumor therapy. This review provides the current overview of the connections between miRNAs and regulation of NK cell functions and discusses the potential of these miRNAs as innovative biomarkers/targets for cancer immunotherapy.
Collapse
Affiliation(s)
- Silvia Pesce
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Marco Greppi
- Department of Experimental Medicine, University of Genoa, Genoa, Italy.,Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Elisa Ferretti
- Department of Experimental Medicine, University of Genoa, Genoa, Italy.,Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Valentina Obino
- Department of Experimental Medicine, University of Genoa, Genoa, Italy.,Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Simona Carlomagno
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Mariangela Rutigliani
- Histological and Anatomical Pathology Unit, Department of Laboratory and Service, E.O. Galliera Hospital, Genova, Italy
| | - Fredrik B Thoren
- Tumor Immunology Laboratory (TIMM) Laboratory at Sahlgrenska Cancer Center, Department of Infectious Diseases, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Simona Sivori
- Department of Experimental Medicine, University of Genoa, Genoa, Italy.,Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | | | - Simona Candiani
- Department of Earth Science, Environment and Life (DISTAV), University of Genoa, Genoa, Italy
| | - Emanuela Marcenaro
- Department of Experimental Medicine, University of Genoa, Genoa, Italy.,Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| |
Collapse
|
12
|
Agarwal S, Kraus Z, Dement-Brown J, Alabi O, Starost K, Tolnay M. Human Fc Receptor-like 3 Inhibits Regulatory T Cell Function and Binds Secretory IgA. Cell Rep 2020; 30:1292-1299.e3. [DOI: 10.1016/j.celrep.2019.12.099] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 12/18/2019] [Accepted: 12/27/2019] [Indexed: 12/17/2022] Open
|
13
|
Shah NM, Edey LF, Imami N, Johnson MR. Human labour is associated with altered regulatory T cell function and maternal immune activation. Clin Exp Immunol 2019; 199:182-200. [PMID: 31617583 DOI: 10.1111/cei.13384] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2019] [Indexed: 12/14/2022] Open
Abstract
During human pregnancy, regulatory T cell (Treg ) function is enhanced and immune activation is repressed allowing the growth and development of the feto-placental unit. Here, we have investigated whether human labour is associated with a reversal of the pregnancy-induced changes in the maternal immune system. We tested the hypothesis that human labour is associated with a decline in Treg function, specifically their ability to modulate Toll-like receptor (TLR)-induced immune responses. We studied the changes in cell number, activation status and functional behaviour of peripheral blood, myometrial (myoMC) and cord blood mononuclear cells (CBMC) with the onset of labour. We found that Treg function declines and that Treg cellular targets change with labour onset. The changes in Treg function were associated with increased activation of myoMC, assessed by their expression of major histocompatibility complex (MHC) class II molecules and CBMC inflammatory cells. The innate immune system showed increased activation, as shown by altered monocyte and neutrophil cell phenotypes, possibly to be ready to respond to microbial invasion after birth or to contribute to tissue remodelling. Our results highlight changes in the function of the adaptive and innate immune systems that may have important roles in the onset of human labour.
Collapse
Affiliation(s)
- N M Shah
- Department of Surgery and Cancer, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - L F Edey
- Department of Surgery and Cancer, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - N Imami
- Department of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, UK
| | - M R Johnson
- Department of Surgery and Cancer, Imperial College London, Chelsea and Westminster Hospital, London, UK
| |
Collapse
|
14
|
Dias ASO, Sacramento PM, Lopes LM, Sales MC, Castro C, Araújo ACRA, Ornelas AMM, Aguiar RS, Silva-Filho RG, Alvarenga R, Bento CAM. TLR-2 and TLR-4 agonists favor expansion of CD4 + T cell subsets implicated in the severity of neuromyelitis optica spectrum disorders. Mult Scler Relat Disord 2019; 34:66-76. [PMID: 31229737 DOI: 10.1016/j.msard.2019.06.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 06/13/2019] [Accepted: 06/16/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND High frequency of circulating Th17 cell subsets expressing TLR2, TLR4 and TLR9 was observed in Neuromyelitis optica spectrum disorder (NMOSD) patients, a severe humoral autoimmune disease of the central nervous system. Our objective was to evaluate the direct effects of different TLR ligands on CD4+ T-cells form those patients. METHODS CD4+ T-cell cultures from NMOSD and healthy individuals were stimulated with different TLR ligands and the cell proliferation and cytokine profile was analyzed by [3H] TdR up take and ELISA/ cytometry, respectively. The plasma levels of CD14 were determined by ELISA. RESULTS Here, Pam3C (TLR2) and LPS (TLR4) induced significant cell proliferation and IL-6, IL-17 and IL-21 production by CD4+ T-cells from NMOSD. Additionally, while both TLR ligands were more potent in favoring the expansion of TFH-like cells, Pam3C reduced the frequency of IL-10-secreting FoxP3+and FoxP3- CD4+ T-cells. With regard to disease severity, the levels of IL-6, IL-17 and IL-21 produced by CD4+ T-cells, as well as the frequency of TFH-like cells, in response to TLR2 and TLR4 agonists were positively correlated with neurological disabilities and the occurrence of new acute relapses during follow up. Finally, circulating levels of CD14, an indirect marker of microbial translocation, were positively correlated with IL-6, IL-17 and IL-21 release by Pam3C- and LPS-activated CD4+ T-cells. CONCLUSIONS In summary, our data suggest that microbial antigens may affect NMOSD outcomes by favoring an imbalance between Th17 and TFH-like cells and regulatory T cell subsets.
Collapse
Affiliation(s)
- Aleida S O Dias
- Department of Microbiology and Parasitology /Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil; Department of Microbiology, Immunology and Parasitology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Priscila M Sacramento
- Department of Microbiology and Parasitology /Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil; Department of Microbiology, Immunology and Parasitology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Lana Márcia Lopes
- Department of Microbiology and Parasitology /Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil; Department of Microbiology, Immunology and Parasitology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Marisa C Sales
- Department of Microbiology, Immunology and Parasitology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Camilla Castro
- Department of Microbiology and Parasitology /Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Carolina R A Araújo
- Departament of Neurology/ Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alice M M Ornelas
- Departament of Genetics, Federal University of Rio de Janeiro, Brazil, Rio de Janeiro, of Rio de Janeiro
| | - Renato S Aguiar
- Departament of Genetics, Federal University of Rio de Janeiro, Brazil, Rio de Janeiro, of Rio de Janeiro
| | - Renato Geraldo Silva-Filho
- Department of Microbiology and Parasitology /Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Regina Alvarenga
- Departament of Neurology/ Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cleonice A M Bento
- Department of Microbiology and Parasitology /Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil; Department of Microbiology, Immunology and Parasitology, Rio de Janeiro State University, Rio de Janeiro, Brazil; Departament of Neurology/ Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
15
|
Targeting IRAK4 disrupts inflammatory pathways and delays tumor development in chronic lymphocytic leukemia. Leukemia 2019; 34:100-114. [PMID: 31197259 PMCID: PMC8075947 DOI: 10.1038/s41375-019-0507-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 03/04/2019] [Accepted: 04/26/2019] [Indexed: 12/20/2022]
Abstract
Interleukin-1 receptor-associated kinase 4 (IRAK4) plays a critical role in Toll-like receptor (TLR) signal transduction and innate immune responses. Recruitment and subsequent activation of IRAK4 upon TLR stimulation is mediated by the myeloid differentiation primary response 88 (MYD88) adaptor protein. Around 3% of chronic lymphocytic leukemia (CLL) patients have activating mutations of MYD88, a driver mutation in this disease. Here, we studied the effects of TLR activation and the pharmacological inhibition of IRAK4 with ND2158, an IRAK4 competitive inhibitor, as a therapeutic approach in CLL. Our in vitro studies demonstrated that ND2158 preferentially killed CLL cells in a dose-dependent manner. We further observed a decrease in NF-κB and STAT3 signaling, cytokine secretion, proliferation and migration of primary CLL cells from MYD88-mutated and -unmutated cases. In the Eµ-TCL1 adoptive transfer mouse model of CLL, ND2158 delayed tumor progression and modulated the activity of myeloid and T cells. Our findings show the importance of TLR signaling in CLL development and suggest IRAK4 as a therapeutic target for this disease.
Collapse
|
16
|
The Regulatory Roles of Toll-Like Receptor 4 in Secretions of Type 1/Type 2 Relative Cytokines by Splenocytes and Dendritic Cells Exposed to Clonorchis sinensis Excretory/Secretory Products. Inflammation 2018; 41:213-220. [PMID: 29047038 DOI: 10.1007/s10753-017-0679-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The roles of TLR4 in mediation of innate immune response and in regulation of adaptive immune responses triggered by Clonorchis sinensis remain unknown. In the present study, splenocytes derived from C3H/HeN (TLR4 wild ) and C3H/Hej mice (TLR4 mut ) that were infected with 45 metacercariae of C. sinensis were harvested, then stimulated by C. sinensis excretory/secretory products (ESP) or medium (control) for 48 h, respectively. Meanwhile, bone marrow-derived dendritic cells (BMDCs) from normal C3H/HeN and C3H/Hej mice were prepared and stimulated with medium, ESP, LPS, or ESP+LPS for 24 h, respectively. The supernatants were collected, and the concentrations of type 1 and type 2 relative cytokines were determined by ELISA. The maturation of BMDCs indicated by surface markers of CD80, CD86, and MHC II was evaluated by flow cytometry. The results showed that the levels of IFN-γ, IL-6, TNF-α, and IL-10 in the splenocytes from C. sinensis-infected TLR4 mut mice were significantly lower than those from TLR4 wild mice when they were further exposed to ESP. For BMDCs, the productions of the cytokines IL-12p70 and IL-10, but not IL-4, in the BMDCs from TLR4 mutation mice were predominantly decreased compared with those from TLR4 wild mice when the BMDCs were co-stimulated by ESP combined with LPS. Flow cytometry analysis showed that ESP could significantly decrease the high levels of CD80, CD86, and MHC II which were elevated by LPS. In conclusion, these data suggest that TLR4 may play a regulatory role in type 1 immune responses during C. sinensis infection.
Collapse
|
17
|
Wang D, Xie N, Gao W, Kang R, Tang D. The ferroptosis inducer erastin promotes proliferation and differentiation in human peripheral blood mononuclear cells. Biochem Biophys Res Commun 2018; 503:1689-1695. [PMID: 30049441 PMCID: PMC6179365 DOI: 10.1016/j.bbrc.2018.07.100] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 07/20/2018] [Indexed: 01/18/2023]
Abstract
Peripheral blood mononuclear cells (PBMCs) contain multipotent progenitor cell populations and possess the potential to differentiate into various types of immune cells under both physiological and pathological conditions. Ferroptosis is a type of oxidative stress-associated cell death that is mainly mediated by lipid peroxidation. However, the function of ferroptosis in cell differentiation remains unknown. Here, we showed that the ferroptosis inducer erastin did not cause cell death in human PBMCs. In contrast, erastin-induced lipid peroxidation promoted human PBMC proliferation and differentiation into B cells and natural killer cells through inhibition of bone morphogenetic protein family expression. These findings uncover a new immune modulation function of erastin in promoting PBMC proliferation and differentiation.
Collapse
Affiliation(s)
- Ding Wang
- The Third Affiliated Hospital, Center for DAMP Biology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Protein Modification and Degradation of Guangdong Higher Education Institutes, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 510510, PR China.
| | - Nan Xie
- Department of Oral Pathology, Guanghua School of Stomatology, Research Institute of Stomatology, Guangdong Province Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong 510055, PR China
| | - Wanli Gao
- The Third Affiliated Hospital, Center for DAMP Biology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Protein Modification and Degradation of Guangdong Higher Education Institutes, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 510510, PR China
| | - Rui Kang
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Daolin Tang
- The Third Affiliated Hospital, Center for DAMP Biology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Protein Modification and Degradation of Guangdong Higher Education Institutes, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, 510510, PR China; Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA.
| |
Collapse
|
18
|
Bavananthasivam J, Alkie TN, Astill J, Abdul-Careem MF, Wootton SK, Behboudi S, Yitbarek A, Sharif S. In ovo administration of Toll-like receptor ligands encapsulated in PLGA nanoparticles impede tumor development in chickens infected with Marek's disease virus. Vaccine 2018; 36:4070-4076. [PMID: 29859800 DOI: 10.1016/j.vaccine.2018.05.091] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 04/20/2018] [Accepted: 05/21/2018] [Indexed: 01/17/2023]
Abstract
One of the economically important diseases in the poultry industry is Marek's disease (MD) which is caused by Marek's disease virus (MDV). The use of current vaccines provides protection against clinical signs of MD in chickens. However, these vaccines do not prevent the transmission of MDV to susceptible hosts, hence they may promote the development of new virulent strains of MDV. This issue persuaded us to explore alternative approaches to control MD in chickens. Induction of innate responses at the early stage of life in the chicken may help to prevent or reduce MDV infection. Further, prophylactic use of Toll-like receptor ligands (TLR-Ls) has been shown to generate host immunity against infectious diseases. In this regard, encapsulation of TLR-Ls in Poly(d, l-lactic-co-glycolic acid) (PLGA) may further enhance host responses by controlled release of TLR-Ls for an extended period. Hence, in the current study, protective effects of encapsulated TLR4 and TLR21 ligands, LPS and CpG, respectively, were investigated against MD. Results indicated that administration of encapsulated CpG and LPS first at embryonic day (ED) 18, followed by post-hatch at 14 days-post infection (dpi) intramuscularly, diminished tumor incidence by 60% and 42.8%, respectively at 21dpi compared to the MDV only group. In addition, analysis of cytokine gene profiles of interferon (IFN)-α, IFN-β, IFN-γ, inducible nitric oxide synthase (iNOS), interleukin (IL)-1β, IL-18 and IL-10 in spleen and bursa of Fabricius at different time points suggests that TLR-Ls possibly triggered host responses through the expression of IL-1β and IL-18 to reduce tumor formation. However, further studies are needed to explore the role of these pro-inflammatory cytokines and other influencing elements like lymphocytes in the hindrance of tumor development by TLR-Ls treatment in chickens.
Collapse
Affiliation(s)
- Jegarubee Bavananthasivam
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Tamiru N Alkie
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Jake Astill
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Mohamed Faizal Abdul-Careem
- Department of Ecosystem and Public Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Sarah K Wootton
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Shahriar Behboudi
- The Pirbright Institute, Ash Road, Pirbright, Woking, Surrey GU24 0NF, United Kingdom; Department of Pathology and Infectious Disease, School of Veterinary Medicine, University of Surrey, Guildford, United Kingdom
| | - Alexander Yitbarek
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Shayan Sharif
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario N1G 2W1, Canada.
| |
Collapse
|
19
|
Ferreira TB, Hygino J, Wing AC, Kasahara TM, Sacramento PM, Camargo S, Rueda F, Alves-Leon SV, Alvarenga R, Vasconcelos CC, Agrawal A, Gupta S, Bento CAM. Different interleukin-17-secreting Toll-like receptor + T-cell subsets are associated with disease activity in multiple sclerosis. Immunology 2017; 154:239-252. [PMID: 29168181 DOI: 10.1111/imm.12872] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 11/04/2017] [Accepted: 11/14/2017] [Indexed: 12/12/2022] Open
Abstract
Signalling through Toll-like receptors (TLRs) may play a role in the pathogenesis of autoimmune diseases, such as multiple sclerosis (MS). In the present study, the expression of TLR-2, -4 and -9 was significantly higher on CD4+ and CD8+ T-cells from MS patients compared to healthy individuals. Following in-vitro activation, the proportion of interleukin (IL)-17+ and IL-6+ CD4+ and CD8+ T-cells was higher in the patients. In addition, the proportion of IFN-γ-secreting TLR+ CD8+ T-cells was increased in MS patients. Among different IL-17+ T-cell phenotypes, the proportion of IL-17+ TLR+ CD4+ and CD8+ T-cells producing IFN-γ or IL-6 were positively associated with the number of active brain lesions and neurological disabilities. Interestingly, activation of purified CD4+ and CD8+ T-cells with ligands for TLR-2 (Pam3Csk4), TLR-4 [lipopolysaccharide (LPS)] and TLR-9 [oligodeoxynucleotide (ODN)] directly induced cytokine production in MS patients. Among the pathogen-associated molecular patterns (PAMPs), Pam3Csk4 was more potent than other TLR ligands in inducing the production of all proinflammatory cytokines. Furthermore, IL-6, IFN-γ, IL-17 and granulocyte-macrophage colony-stimulating factor (GM-CSF) levels produced by Pam3Csk4-activated CD4+ cells were directly associated with disease activity. A similar correlation was observed with regard to IL-17 levels released by Pam3Csk4-stimulated CD8+ T-cells and clinical parameters. In conclusion, our data suggest that the expansion of different T helper type 17 (Th17) phenotypes expressing TLR-2, -4 and -9 is associated with MS disease activity, and reveals a preferential ability of TLR-2 ligand in directly inducing the production of cytokines related to brains lesions and neurological disabilities.
Collapse
Affiliation(s)
- Thais B Ferreira
- Post-graduate Program in Microbiology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Joana Hygino
- Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Cristina Wing
- Post-graduate Program Neurology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Taissa M Kasahara
- Post-graduate Program in Microbiology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Priscila M Sacramento
- Post-graduate Program in Microbiology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Solange Camargo
- Lagoa Hospital, Barra da Tijuca Unity, Rio de Janeiro, Brazil
| | - Fernanda Rueda
- Clinical of Diagnosis by Image, Barra da Tijuca Unity, Rio de Janeiro, Brazil
| | - Soniza V Alves-Leon
- Post-graduate Program Neurology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Regina Alvarenga
- Post-graduate Program Neurology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | - Cleonice A M Bento
- Post-graduate Program in Microbiology, University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.,Department of Microbiology and Parasitology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.,Post-graduate Program Neurology, Federal University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
20
|
Merlini E, Tincati C, Biasin M, Saulle I, Cazzaniga FA, d'Arminio Monforte A, Cappione AJ, Snyder-Cappione J, Clerici M, Marchetti GC. Stimulation of PBMC and Monocyte-Derived Macrophages via Toll-Like Receptor Activates Innate Immune Pathways in HIV-Infected Patients on Virally Suppressive Combination Antiretroviral Therapy. Front Immunol 2016; 7:614. [PMID: 28066424 PMCID: PMC5165253 DOI: 10.3389/fimmu.2016.00614] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 12/05/2016] [Indexed: 12/25/2022] Open
Abstract
In HIV-infected, combination antiretroviral therapy (cART)-treated patients, immune activation and microbial translocation persist and associate with inadequate CD4 recovery and morbidity/mortality. We analyzed whether alterations in the toll-like receptor (TLR) pathway could be responsible for the immune hyperactivation seen in these patients. PBMC/monocyte-derived macrophages (MDMs) of 28 HIV+ untreated and 35 cART-treated patients with HIV-RNA < 40 cp/mL [20 Full Responders (FRs): CD4 ≥ 350; 15 Immunological Non-Responders (INRs): CD4 < 350], as well as of 16 healthy controls were stimulated with a panel of TLR agonists. We measured: CD4/CD8/CD14/CD38/HLA-DR/Ki67/AnnexinV/CD69/TLR4/8 (Flow Cytometry); PBMC expression of 84 TLR pathway genes (qPCR); PBMC/MDM cytokine release (Multiplex); and plasma lipopolysaccharide (LPS)/sCD14 (LAL/ELISA). PBMC/MDM from cART patients responded weakly to LPS stimulation but released high amounts of pro-inflammatory cytokines. MDM from these patients were characterized by a reduced expression of HLA-DR+ MDM and failed to expand activated HLA-DR+ CD38+ T-lymphocytes. PBMC/MDM from cART patients responded more robustly to ssRNA stimulation; this resulted in a significant expansion of activated CD38 + CD8 and the release of amounts of pro-inflammatory cytokines comparable to those seen in untreated viremic patients. Despite greater constitutive TLR pathway gene expression, PBMC from INRs seemed to upregulate only type I IFN genes following TLR stimulation, whereas PBMC from full responders showed a broader response. Systemic exposure to microbial antigens drives immune activation during cART by triggering TLRs. Bacterial stimulation modifies MDM function/pro-inflammatory profile in cART patients without affecting T-lymphocytes; this suggests translocating bacteria as selective stimulus to chronic innate activation during cART. High constitutive TLR activation is seen in patients lacking CD4 recovery, suggesting an exhausted immune milieu, anergic to further antigen encounters.
Collapse
Affiliation(s)
- Esther Merlini
- Department of Health Sciences, Clinic of Infectious Diseases, ASST Santi Paolo e Carlo, University of Milan , Milan , Italy
| | - Camilla Tincati
- Department of Health Sciences, Clinic of Infectious Diseases, ASST Santi Paolo e Carlo, University of Milan , Milan , Italy
| | - Mara Biasin
- Department of Biomedical and Clinical Sciences - "L. Sacco", University of Milan , Milan , Italy
| | - Irma Saulle
- Department of Biomedical and Clinical Sciences - "L. Sacco", University of Milan , Milan , Italy
| | - Federico Angelo Cazzaniga
- Department of Health Sciences, Clinic of Infectious Diseases, ASST Santi Paolo e Carlo, University of Milan , Milan , Italy
| | - Antonella d'Arminio Monforte
- Department of Health Sciences, Clinic of Infectious Diseases, ASST Santi Paolo e Carlo, University of Milan , Milan , Italy
| | | | | | - Mario Clerici
- Department of Physiopathology and Transplants, University of Milan, Milan, Italy; Don C. Gnocchi Foundation, Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Giulia Carla Marchetti
- Department of Health Sciences, Clinic of Infectious Diseases, ASST Santi Paolo e Carlo, University of Milan , Milan , Italy
| |
Collapse
|
21
|
Yong MK, Cameron PU, Spelman T, Elliott JH, Fairley CK, Boyle J, Miyamasu M, Lewin SR. Quantifying Adaptive and Innate Immune Responses in HIV-Infected Participants Using a Novel High Throughput Assay. PLoS One 2016; 11:e0166549. [PMID: 27935986 PMCID: PMC5147821 DOI: 10.1371/journal.pone.0166549] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 10/31/2016] [Indexed: 11/19/2022] Open
Abstract
Objectives HIV infection is characterised by persistent immune dysfunction of both the adaptive and innate immune responses. The aim of this study was to evaluate these responses using a novel high throughput assay in healthy controls and HIV-infected individuals prior to and following anti-retroviral treatment (ART). Design Cross-sectional study. Methods Whole blood was assessed using the QuantiFERON Monitor® (QFM) assay containing adaptive and innate immunostimulants. Interferon (IFN)-γ levels (IU/mL) were measured by enzyme-linked immunosorbent assay (ELISA). Results We recruited HIV-infected participants (n = 20 off ART and viremic; n = 59 on suppressive ART) and HIV-uninfected controls (n = 229). Median IFN-γ production was significantly higher in HIV-infected participants compared to controls (IFN-γ 512 vs 223 IU/ml, p<0.0001), but within the HIV-infected participants there was no difference between those on or off ART (median IFN-γ 512 vs 593 IU/ml p = 0.94). Amongst the HIV-infected participants, IFN-γ production was higher in individuals with CD4 count>350 compared to <350 cells/μL (IFN-γ IU/ml 561 vs 259 p = 0.02) and in males compared to females (IFN-γ 542 vs 77 IU/ml p = 0.04). There were no associations between IFN-γ production and age, plasma HIV RNA, nadir CD4 count or duration of HIV infection. Using a multivariable analysis, neither CD4 nor sex were independently predictive of IFN-γ production. Conclusion Using a high throughput assay which assesses both adaptive and innate immune function, we showed elevated IFN-γ production in HIV-infected patients both on and off ART. Further research is warranted to determine if changes in QuantiFERON Monitor® are associated with clinical outcomes.
Collapse
Affiliation(s)
- Michelle K. Yong
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Australia
- Department of Infectious Diseases, Monash University and Alfred Hospital, Melbourne, Australia
- * E-mail:
| | - Paul U. Cameron
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Australia
- Department of Infectious Diseases, Monash University and Alfred Hospital, Melbourne, Australia
| | - Tim Spelman
- Centre for Population Health, Burnet Institute, Melbourne, Australia
| | - Julian H. Elliott
- Department of Infectious Diseases, Monash University and Alfred Hospital, Melbourne, Australia
| | - Christopher K. Fairley
- Central Clinical School, Monash University and Melbourne Sexual Health Centre, Melbourne, Australia
| | - Jeffrey Boyle
- QIAGEN, Germantown, Maryland, United States of America
| | | | - Sharon R. Lewin
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Australia
- Department of Infectious Diseases, Monash University and Alfred Hospital, Melbourne, Australia
| |
Collapse
|
22
|
Gerriets VA, Kishton RJ, Johnson MO, Cohen S, Siska PJ, Nichols AG, Warmoes MO, de Cubas AA, MacIver NJ, Locasale JW, Turka LA, Wells AD, Rathmell JC. Foxp3 and Toll-like receptor signaling balance T reg cell anabolic metabolism for suppression. Nat Immunol 2016; 17:1459-1466. [PMID: 27695003 DOI: 10.1038/ni.3577] [Citation(s) in RCA: 409] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 09/02/2016] [Indexed: 02/07/2023]
Abstract
CD4+ effector T cells (Teff cells) and regulatory T cells (Treg cells) undergo metabolic reprogramming to support proliferation and immunological function. Although signaling via the lipid kinase PI(3)K (phosphatidylinositol-3-OH kinase), the serine-threonine kinase Akt and the metabolic checkpoint kinase complex mTORC1 induces both expression of the glucose transporter Glut1 and aerobic glycolysis for Teff cell proliferation and inflammatory function, the mechanisms that regulate Treg cell metabolism and function remain unclear. We found that Toll-like receptor (TLR) signals that promote Treg cell proliferation increased PI(3)K-Akt-mTORC1 signaling, glycolysis and expression of Glut1. However, TLR-induced mTORC1 signaling also impaired Treg cell suppressive capacity. Conversely, the transcription factor Foxp3 opposed PI(3)K-Akt-mTORC1 signaling to diminish glycolysis and anabolic metabolism while increasing oxidative and catabolic metabolism. Notably, Glut1 expression was sufficient to increase the number of Treg cells, but it reduced their suppressive capacity and Foxp3 expression. Thus, inflammatory signals and Foxp3 balance mTORC1 signaling and glucose metabolism to control the proliferation and suppressive function of Treg cells.
Collapse
Affiliation(s)
- Valerie A Gerriets
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, USA
| | - Rigel J Kishton
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, USA
| | - Marc O Johnson
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, USA.,Department of Pathology, Microbiology, and Immunology, Vanderbilt Center for Immunobiology, Vanderbilt University, Nashville, Tennessee, USA
| | - Sivan Cohen
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, USA
| | - Peter J Siska
- Department of Pathology, Microbiology, and Immunology, Vanderbilt Center for Immunobiology, Vanderbilt University, Nashville, Tennessee, USA
| | - Amanda G Nichols
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, USA
| | - Marc O Warmoes
- Center for Environmental and Systems Biochemistry, Department of Toxicology and Cancer Biology and Markey Cancer Center, University of Kentucky, Lexington, Kentucky, USA
| | - Aguirre A de Cubas
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Nancie J MacIver
- Division of Pediatric Endocrinology and Diabetes, Duke University, Durham, North Carolina, USA
| | - Jason W Locasale
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, USA
| | - Laurence A Turka
- Massachusetts General Hospital, Center for Transplantation Sciences, Boston, Massachusetts, USA
| | - Andrew D Wells
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jeffrey C Rathmell
- Department of Pathology, Microbiology, and Immunology, Vanderbilt Center for Immunobiology, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
23
|
Grumelli S. Choline Triggers Exacerbations of Chronic Obstructive Pulmonary Disease in Patients Infected with Pseudomonas aeruginosa. CURRENT RESPIRATORY MEDICINE REVIEWS 2016; 12:167-174. [PMID: 29386986 DOI: 10.2174/1573398x12999160506104327] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background Although exacerbations of chronic obstructive pulmonary disease produced by Pseudomonas aeruginosa infections are a major cause of death, the molecular mechanism that produces them is not well known. Here we focused on the energetic basis of dyspnoea, hypercapnia and acidosis symptoms. Methods and Findings We used an in vivo exacerbation model exposing mice to cigarette smoke and LPS, to mimic emphysema and infections, and choline challenges to trigger exacerbations, that showed 31% increased in the airway resistance for naïve mice and 250% for smoke/LPS treatment. Tissue resistance was increased 32%, in naïve mice, and 169% for smoke/LPS treatment. A decreased tissue elastance, was confirmed by decreased collagen content and increased alveoli chord length. Consequently, the O2 demanded was 260% greater for smoke/LPS treated mice, to provide the energy required to pump the same volume of air then for naïve mice. The extra CO2 produced per ml of air pumped caused hypercapnia and acidosis by 4% decrease in pH.In addition, the bacteria grown with choline had a decrease of 67% in phosphate, 23% ATP and 85% phospholipids with an increase of 57% in polyphosphates, 50% carbohydrates, 100% LPS, consuming 45% less energy relative to the bacteria grown with succinate. Conclusion choline, released by P. aeruginosa, triggers exacerbation symptoms by increasing lung resistance, O2 consumption and producing more pCO2 in blood with dyspnea, hypercapnia and acidosis. The energetic shift of decreased O2 bacterial demand and increased lung demand benefits the infection, thus restoring the energetic balance on the host will favor P. aeruginosa eradication.
Collapse
Affiliation(s)
- Sandra Grumelli
- Centro de Investigaciones en Medicina Respiratoria, Universidad Católica de Córdoba, Cordoba, Argentina.,Pulmonary Division of the Brigham and Women's Hospital, Harvard Medical School, Boston, USA.,Departamento de Biologia Molecular, Universidad Nacional de Rio Cuarto, Rio Cuarto, Córdoba, Argentina
| |
Collapse
|
24
|
Gong Y, Tao L, Jing L, Liu D, Hu S, Liu W, Zhou N, Xie Y. Association of TLR4 and Treg in Helicobacter pylori Colonization and Inflammation in Mice. PLoS One 2016; 11:e0149629. [PMID: 26901645 PMCID: PMC4762684 DOI: 10.1371/journal.pone.0149629] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 02/03/2016] [Indexed: 12/14/2022] Open
Abstract
The host immune response plays an important role in the pathogenesis of Helicobacter pylori infection. The aim of this study was to clarify the immune pathogenic mechanism of Helicobacter pylori infection via TLR signaling and gastric mucosal Treg cells in mice. To discover the underlying mechanism, we selectively blocked the TLR signaling pathway and subpopulations of regulatory T cells in the gastric mucosa of mice, and examined the consequences on H. pylori infection and inflammatory response as measured by MyD88, NF-κB p65, and Foxp3 protein expression levels and the levels of Th1, Th17 and Th2 cytokines in the gastric mucosa. We determined that blocking TLR4 signaling in H. pylori infected mice decreased the numbers of Th1 and Th17 Treg cells compared to controls (P < 0.001-0.05), depressed the immune response as measured by inflammatory grade (P < 0.05), and enhanced H. pylori colonization (P < 0.05). In contrast, blocking CD25 had the opposite effects, wherein the Th1 and Th17 cell numbers were increased (P < 0.001-0.05), immune response was enhanced (P < 0.05), and H. pylori colonization was inhibited (P < 0.05) compared to the non-blocked group. In both blocked groups, the Th2 cytokine IL-4 remained unchanged, although IL-10 in the CD25 blocked group was significantly decreased (P < 0.05). Furthermore, MyD88, NF-κB p65, and Foxp3 in the non-blocked group were significantly lower than those in the TLR4 blocked group (P < 0.05), but significantly higher than those of the CD25 blocked group (P < 0.05). Together, these results suggest that there might be an interaction between TLR signaling and Treg cells that is important for limiting H. pylori colonization and suppressing the inflammatory response of infected mice.
Collapse
Affiliation(s)
- Yanfeng Gong
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi, China
- Department of Geriatrics, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi, China
| | - Liming Tao
- Department of Obstetrics, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi, China
| | - Lei Jing
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi, China
| | - Dongsheng Liu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi, China
| | - Sijun Hu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi, China
| | - Wei Liu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi, China
| | - Nanjin Zhou
- Department of Biochemistry and Molecular Biology, Jiangxi Academy of Medical Science, Jiangxi, China
- * E-mail: (YX); (NZ)
| | - Yong Xie
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, No. 17, Yongwai Zheng Street, Nanchang, Jiangxi, China
- * E-mail: (YX); (NZ)
| |
Collapse
|
25
|
Kou F, Lu Z, Li J, Zhang X, Lu M, Zhou J, Wang X, Gong J, Gao J, Li J, Li Y, Shen L. Pretreatment lymphopenia is an easily detectable predictive and prognostic marker in patients with metastatic esophagus squamous cell carcinoma receiving first-line chemotherapy. Cancer Med 2016; 5:778-86. [PMID: 26814381 PMCID: PMC4864807 DOI: 10.1002/cam4.638] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 11/22/2015] [Accepted: 12/16/2015] [Indexed: 01/21/2023] Open
Abstract
To explore the influence of pretreatment lymphopenia on the toxicity and efficacy of first‐line chemotherapy in patients with metastatic esophagus squamous cell carcinoma (ESCC). In total, 215 patients were included in this retrospective study. Correlations between pretreatment lymphopenia (lymphocyte count <1 × 109/L) and the occurrence of toxicity and the efficacy of first‐line palliative chemotherapy were investigated. Pretreatment lymphopenia was found in 19.1% of the patients. The overall response rate (ORR) was 35.5% (65 of 183 patients). Patients with pretreatment lymphopenia had a lower ORR to chemotherapy compared with those without lymphopenia (22.2% vs. 38.8%, respectively; P = 0.045). Furthermore, the patients with pretreatment lymphopenia have higher grade 3–4 hematological toxicity than that of patients without pretreatment lymphopenia (19 of 41 patients, 46.3% vs. 54 of 174 patients, 31.0%; P = 0.048). Pretreatment lymphopenia was not correlated with grade 3–4 nonhematological toxicity. Multivariate analysis showed that pretreatment lymphopenia is an independent prognostic factor. Patients with pretreatment lymphopenia had a significantly shorter overall survival time than those without lymphopenia (8.2 months vs. 12.7 months; P = 0.020). This study shows that pretreatment lymphopenia is a good prognostic factor as well as a predictive factor for tumor response and chemotherapy‐related hematological toxicity in metastatic ESCC.
Collapse
Affiliation(s)
- Furong Kou
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of GI Oncology, Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing, 100142, China
| | - Zhihao Lu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of GI Oncology, Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing, 100142, China
| | - Jian Li
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of GI Oncology, Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing, 100142, China
| | - Xiaotian Zhang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of GI Oncology, Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing, 100142, China
| | - Ming Lu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of GI Oncology, Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing, 100142, China
| | - Jun Zhou
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of GI Oncology, Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing, 100142, China
| | - Xicheng Wang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of GI Oncology, Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing, 100142, China
| | - Jifang Gong
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of GI Oncology, Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing, 100142, China
| | - Jing Gao
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of GI Oncology, Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing, 100142, China
| | - Jie Li
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of GI Oncology, Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing, 100142, China
| | - Yan Li
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of GI Oncology, Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing, 100142, China
| | - Lin Shen
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of GI Oncology, Peking University School of Oncology, Beijing Cancer Hospital & Institute, Beijing, 100142, China
| |
Collapse
|
26
|
The TLR7 agonist induces tumor regression both by promoting CD4⁺T cells proliferation and by reversing T regulatory cell-mediated suppression via dendritic cells. Oncotarget 2015; 6:1779-89. [PMID: 25593198 PMCID: PMC4359331 DOI: 10.18632/oncotarget.2757] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 11/16/2014] [Indexed: 11/25/2022] Open
Abstract
Treg-induced immunosuppression is now recognized as a key element in enabling tumors to escape immune-mediated destruction. Although topical TLR7 therapies such as imiquimod have been proved successful in the treatment of dermatological malignancy and a number of conditions beyond the FDA-approved indications, the mechanism behind the effect of TLR7 on effector T cell and Treg cell function in cancer immunosurveillance is still not well understood. Here, we found that Loxoribin, one of the TLR7 ligands, could inhibit tumor growth in xenograft models of colon cancer and lung cancer, and these anti-tumor effects of Loxoribin were mediated by promoting CD4⁺T cell proliferation and reversing Treg-mediated suppression via dendritic cells (DCs). However, deprivation of IL-6 using a neutralizing antibody abrogated the ability of Loxoribin-treated DCs, which reversed the Treg cell-mediated suppression. Furthermore, adoptive transfer of Loxoribin-treated DCs inhibited the tumor growth in vivo. Thus, this study links TLR7 signaling to the functional control of effector T cells and Treg cells and identifies Loxoribin as a new therapeutic strategy in cancer treatment, which may offer new opportunities to improve the outcome of cancer immunotherapy.
Collapse
|
27
|
Jasiulewicz A, Lisowska KA, Pietruczuk K, Frąckowiak J, Fulop T, Witkowski JM. Homeostatic 'bystander' proliferation of human peripheral blood B cells in response to polyclonal T-cell stimulation in vitro. Int Immunol 2015; 27:579-88. [PMID: 25995267 DOI: 10.1093/intimm/dxv032] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 05/15/2015] [Indexed: 12/15/2022] Open
Abstract
The mechanisms of maintenance of adequate numbers of B lymphocytes and of protective levels of immunoglobulins in the absence of antigenic (re)stimulation remain not fully understood. Meanwhile, our results presented here show that both peripheral blood naive and memory B cells can be activated strongly and non-specifically (in a mitogen-like fashion) in 5-day in vitro cultures of anti-CD3- or concanavalin A (Con A)-stimulated peripheral blood mononuclear cells of healthy people. This polyclonal, bystander activation of the B cells includes multiple divisions of most of them (assessed here by the flow cytometric technique of dividing cell tracking) and significant antibody [immunoglobulin M (IgM) and IgG] secretion. Observed proliferation of the CD19(+) B cells depends on contact with stimulated T helper (Th) cells (via CD40-CD40L interaction) and on the response of B cells to secreted interleukins IL-5, IL-10 and IL-4, and is correlated with the levels of these Th-derived molecules, while it does not involve the ligation of the BCR/CD19 complex. We suggest that the effect might reflect the situation occurring in vivo as the homeostatic proliferation of otherwise non-stimulated, peripheral B lymphocytes, providing an always ready pool for efficient antibody production to any new (or cognate) antigen challenge.
Collapse
Affiliation(s)
- Aleksandra Jasiulewicz
- Department of Pathophysiology, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| | - Katarzyna A Lisowska
- Department of Pathophysiology, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| | - Krzysztof Pietruczuk
- Department of Pathophysiology, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| | - Joanna Frąckowiak
- Department of Pathophysiology, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| | - Tamas Fulop
- Research Center on Aging, Department of Medicine, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Jacek M Witkowski
- Department of Pathophysiology, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland
| |
Collapse
|
28
|
Karimi S, Chattopadhyay S, Chakraborty NG. Manipulation of regulatory T cells and antigen-specific cytotoxic T lymphocyte-based tumour immunotherapy. Immunology 2015; 144:186-96. [PMID: 25243729 DOI: 10.1111/imm.12387] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 09/12/2014] [Accepted: 09/16/2014] [Indexed: 12/16/2022] Open
Abstract
The most potent killing machinery in our immune system is the cytotoxic T lymphocyte (CTL). Since the possibility for self-destruction by these cells is high, many regulatory activities exist to prevent autoimmune destruction by these cells. A tumour (cancer) grows from the cells of the body and is tolerated by the body's immune system. Yet, it has been possible to generate tumour-associated antigen (TAA) -specific CTL that are also self-antigen specific in vivo, to achieve a degree of therapeutic efficacy. Tumour-associated antigen-specific T-cell tolerance through pathways of self-tolerance generation represents a significant challenge to successful immunotherapy. CD4(+) CD25(+) FoxP3(+) T cells, referred to as T regulatory (Treg) cells, are selected in the thymus as controllers of the anti-self repertoire. These cells are referred to as natural T regulatory (nTreg) cells. According to the new consensus (Nature Immunology 2013; 14:307-308) these cells are to be termed as (tTreg). There is another class of CD4(+) Treg cells also involved in regulatory function in the periphery, also phenotypically CD4(+) CD25(±) , classified as induced Treg (iTreg) cells. These cells are to be termed as peripherally induced Treg (pTreg) cells. In vitro-induced Treg cells with suppressor function should be termed as iTreg. These different Treg cells differ in their requirements for activation and in their mode of action. The current challenges are to determine the degree of specificity of these Treg cells in recognizing the same TAA as the CTL population and to circumvent their regulatory constraints so as to achieve robust CTL responses against cancer.
Collapse
Affiliation(s)
- Shirin Karimi
- Department of Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | | | | |
Collapse
|